Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
JCI Insight ; 9(9)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38512434

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an expanded polyglutamine tract in the widely expressed ataxin-1 (ATXN1) protein. To elucidate anatomical regions and cell types that underlie mutant ATXN1-induced disease phenotypes, we developed a floxed conditional knockin mouse (f-ATXN1146Q/2Q) with mouse Atxn1 coding exons replaced by human ATXN1 exons encoding 146 glutamines. f-ATXN1146Q/2Q mice manifested SCA1-like phenotypes including motor and cognitive deficits, wasting, and decreased survival. Central nervous system (CNS) contributions to disease were revealed using f-ATXN1146Q/2Q;Nestin-Cre mice, which showed improved rotarod, open field, and Barnes maze performance by 6-12 weeks of age. In contrast, striatal contributions to motor deficits using f-ATXN1146Q/2Q;Rgs9-Cre mice revealed that mice lacking ATXN1146Q/2Q in striatal medium-spiny neurons showed a trending improvement in rotarod performance at 30 weeks of age. Surprisingly, a prominent role for muscle contributions to disease was revealed in f-ATXN1146Q/2Q;ACTA1-Cre mice based on their recovery from kyphosis and absence of muscle pathology. Collectively, data from the targeted conditional deletion of the expanded allele demonstrated CNS and peripheral contributions to disease and highlighted the need to consider muscle in addition to the brain for optimal SCA1 therapeutics.


Subject(s)
Ataxin-1 , Disease Models, Animal , Muscle, Skeletal , Spinocerebellar Ataxias , Animals , Ataxin-1/genetics , Ataxin-1/metabolism , Mice , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Humans , Male , Mice, Transgenic , Gene Knock-In Techniques , Female , Phenotype , Neurons/metabolism , Neurons/pathology
2.
bioRxiv ; 2023 May 11.
Article in English | MEDLINE | ID: mdl-37214832

ABSTRACT

Spinocerebellar ataxia type 7 (SCA7) is an inherited neurodegenerative disorder caused by a CAG-polyglutamine repeat expansion. SCA7 patients display a striking loss of Purkinje cell (PC) neurons with disease progression; however, PCs are rare, making them difficult to characterize. We developed a PC nuclei enrichment protocol and applied it to single-nucleus RNA-seq of a SCA7 knock-in mouse model. Our results unify prior observations into a central mechanism of cell identity loss, impacting both glia and PCs, driving accumulation of inhibitory synapses and altered PC spiking. Zebrin-II subtype dysregulation is the predominant signal in PCs, leading to complete loss of zebrin-II striping at motor symptom onset in SCA7 mice. We show this zebrin-II subtype degradation is shared across Polyglutamine Ataxia mouse models and SCA7 patients. It has been speculated that PC subtype organization is critical for cerebellar function, and our results suggest that a breakdown of zebrin-II parasagittal striping is pathological.

3.
bioRxiv ; 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-36798410

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an expanded polyglutamine tract in the widely expressed ATXN1 protein. To elucidate anatomical regions and cell types that underlie mutant ATXN1-induced disease phenotypes, we developed a floxed conditional knockout mouse model ( f-ATXN1 146Q/2Q ) having mouse Atxn1 coding exons replaced by human exons encoding 146 glutamines. F-ATXN1 146Q/2Q mice manifest SCA1-like phenotypes including motor and cognitive deficits, wasting, and decreased survival. CNS contributions to disease were revealed using ATXN1 146Q/2Q ; Nestin-Cre mice, that showed improved rotarod, open field and Barnes maze performances. Striatal contributions to motor deficits were examined using f-ATXN1 146Q/2Q ; Rgs9-Cre mice. Mice lacking striatal ATXN1 146Q/2Q had improved rotarod performance late in disease. Muscle contributions to disease were revealed in f-ATXN1 146Q/2Q ; ACTA1-Cre mice which lacked muscle pathology and kyphosis seen in f-ATXN1 146Q/2Q mice. Kyphosis was not improved in f-ATXN1 146Q/2Q ;Nestin - Cre mice. Thus, optimal SCA1 therapeutics will require targeting mutant ATXN1 toxic actions in multiple brain regions and muscle.

4.
Neuron ; 111(4): 493-507.e6, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36577403

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a dominant trinucleotide repeat neurodegenerative disease characterized by motor dysfunction, cognitive impairment, and premature death. Degeneration of cerebellar Purkinje cells is a frequent and prominent pathological feature of SCA1. We previously showed that transport of ATXN1 to Purkinje cell nuclei is required for pathology, where mutant ATXN1 alters transcription. To examine the role of ATXN1 nuclear localization broadly in SCA1-like disease pathogenesis, CRISPR-Cas9 was used to develop a mouse with an amino acid alteration (K772T) in the nuclear localization sequence of the expanded ATXN1 protein. Characterization of these mice indicates that proper nuclear localization of mutant ATXN1 contributes to many disease-like phenotypes including motor dysfunction, cognitive deficits, and premature lethality. RNA sequencing analysis of genes with expression corrected to WT levels in Atxn1175QK772T/2Q mice indicates that transcriptomic aspects of SCA1 pathogenesis differ between the cerebellum, brainstem, cerebral cortex, hippocampus, and striatum.


Subject(s)
Ataxin-1 , Spinocerebellar Ataxias , Transcriptome , Animals , Mice , Ataxin-1/genetics , Ataxin-1/metabolism , Brain/metabolism , Cerebellum/metabolism , Disease Models, Animal , Mice, Transgenic , Nerve Tissue Proteins/genetics , Phenotype , Protein Transport/genetics , Purkinje Cells/metabolism , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism
5.
Mol Ther Nucleic Acids ; 21: 1006-1016, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32818920

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a lethal, autosomal dominant neurodegenerative disease caused by a polyglutamine expansion in the ATAXIN-1 (ATXN1) protein. Preclinical studies demonstrate the therapeutic efficacy of approaches that target and reduce Atxn1 expression in a non-allele-specific manner. However, studies using Atxn1-/- mice raise cautionary notes that therapeutic reductions of ATXN1 might lead to undesirable effects such as reduction in the activity of the tumor suppressor Capicua (CIC), activation of the protease ß-secretase 1 (BACE1) and subsequent increased amyloidogenic cleavage of the amyloid precursor protein (APP), or a reduction in hippocampal neuronal precursor cells that would impact hippocampal function. Here, we tested whether an antisense oligonucleotide (ASO)-mediated reduction of Atxn1 produced unwanted effects involving BACE1, CIC activity, or reduction in hippocampal neuronal precursor cells. Notably, no effects on BACE1, CIC tumor suppressor function, or number of hippocampal neuronal precursor cells were found in mice subjected to a chronic in vivo ASO-mediated reduction of Atxn1. These data provide further support for targeted reductions of ATXN1 as a therapeutic approach for SCA1.

6.
Nat Commun ; 11(1): 3343, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620905

ABSTRACT

The expanded polyglutamine (polyQ) tract form of ataxin-1 drives disease progression in spinocerebellar ataxia type 1 (SCA1). Although known to form distinctive intranuclear bodies, the cellular pathways and processes that polyQ-ataxin-1 influences remain poorly understood. Here we identify the direct and proximal partners constituting the interactome of ataxin-1[85Q] in Neuro-2a cells, pathways analyses indicating a significant enrichment of essential nuclear transporters, pointing to disruptions in nuclear transport processes in the presence of elevated levels of ataxin-1. Our direct assessments of nuclear transporters and their cargoes confirm these observations, revealing disrupted trafficking often with relocalisation of transporters and/or cargoes to ataxin-1[85Q] nuclear bodies. Analogous changes in importin-ß1, nucleoporin 98 and nucleoporin 62 nuclear rim staining are observed in Purkinje cells of ATXN1[82Q] mice. The results highlight a disruption of multiple essential nuclear protein trafficking pathways by polyQ-ataxin-1, a key contribution to furthering understanding of pathogenic mechanisms initiated by polyQ tract proteins.


Subject(s)
Ataxin-1/metabolism , Cell Nucleus/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Purkinje Cells/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Ataxin-1/genetics , Cell Line, Tumor , Disease Models, Animal , HeLa Cells , Humans , Mice , Mutation , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Peptides/genetics , Protein Binding , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism , Trinucleotide Repeat Expansion/genetics
7.
Neuropsychopharmacology ; 45(7): 1159-1170, 2020 06.
Article in English | MEDLINE | ID: mdl-32179875

ABSTRACT

Autism spectrum disorder (ASD) encompasses wide-ranging neuropsychiatric symptoms with unclear etiology. Although the cerebellum is a key region implicated in ASD, it remains elusive how the cerebellar circuitry is altered and whether the cerebellum can serve as a therapeutic target to rectify the phenotype of idiopathic ASD with polygenic abnormalities. Using a syndromic ASD model, e.g., Black and Tan BRachyury T+Itpr3tf/J (BTBR) mice, we revealed that increased excitability of presynaptic interneurons (INs) and decreased intrinsic excitability of postsynaptic Purkinje neurons (PNs) resulted in low PN firing rates in the cerebellum. Knowing that downregulation of Kv1.2 potassium channel in the IN nerve terminals likely augmented their excitability and GABA release, we applied a positive Kv1.2 modulator to mitigate the presynaptic over-inhibition and social impairment of BTBR mice. Selective restoration of the PN activity by a new chemogenetic approach alleviated core ASD-like behaviors of the BTBR strain. These findings highlight complex mechanisms converging onto the cerebellar dysfunction in the phenotypic model and provide effective strategies for potential therapies of ASD.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Cerebellum , Animals , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/genetics , Cerebellum/physiopathology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL
8.
Hum Mol Genet ; 29(1): 117-131, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31696233

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by abnormal expansion of glutamine (Q) encoding CAG repeats in the gene Ataxin-1 (ATXN1). Although motor and balance deficits are the core symptoms of SCA1, cognitive decline is also commonly observed in patients. While mutant ATXN1 is expressed throughout the brain, pathological findings reveal severe atrophy of cerebellar cortex in SCA1 patients. The cerebellum has recently been implicated in diverse cognitive functions, yet to what extent cerebellar neurodegeneration contributes to cognitive alterations in SCA1 remains poorly understood. Much of our understanding of the mechanisms underlying pathogenesis of motor symptoms in SCA1 comes from mouse models. Reasoning that mouse models could similarly offer important insights into the mechanisms of cognitive alterations in SCA1, we tested cognition in several mouse lines using Barnes maze and fear conditioning. We confirmed cognitive deficits in Atxn1154Q/2Q knock-in mice with brain-wide expression of mutant ATXN1 and in ATXN1 null mice. We found that shorter polyQ length and haploinsufficiency of ATXN1 do not cause significant cognitive deficits. Finally, ATXN1[82Q ] transgenic mice-with cerebellum limited expression of mutant ATXN1-demonstrated milder impairment in most aspects of cognition compared to Atxn1154Q/2Q mice, supporting the concept that cognitive deficits in SCA1 arise from a combination of cerebellar and extra-cerebellar dysfunctions.


Subject(s)
Ataxin-1/metabolism , Cerebellum/metabolism , Cognitive Dysfunction/metabolism , Animals , Ataxin-1/genetics , Ataxin-3/genetics , Ataxin-3/metabolism , Cognition/physiology , Cognitive Dysfunction/genetics , Disease Models, Animal , Female , Male , Mice , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism
9.
JCI Insight ; 3(21)2018 11 02.
Article in English | MEDLINE | ID: mdl-30385727

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited ataxia caused by expansion of a translated CAG repeat encoding a glutamine tract in the ataxin-1 (ATXN1) protein. Despite advances in understanding the pathogenesis of SCA1, there are still no therapies to alter its progressive fatal course. RNA-targeting approaches have improved disease symptoms in preclinical rodent models of several neurological diseases. Here, we investigated the therapeutic capability of an antisense oligonucleotide (ASO) targeting mouse Atxn1 in Atxn1154Q/2Q-knockin mice that manifest motor deficits and premature lethality. Following a single ASO treatment at 5 weeks of age, mice demonstrated rescue of these disease-associated phenotypes. RNA-sequencing analysis of genes with expression restored to WT levels in ASO-treated Atxn1154Q/2Q mice was used to demonstrate molecular differences between SCA1 pathogenesis in the cerebellum and disease in the medulla. Finally, select neurochemical abnormalities detected by magnetic resonance spectroscopy in vehicle-treated Atxn1154Q/2Q mice were reversed in the cerebellum and brainstem (a region containing the pons and the medulla) of ASO-treated Atxn1154Q/2Q mice. Together, these findings support the efficacy and therapeutic importance of directly targeting ATXN1 RNA expression as a strategy for treating both motor deficits and lethality in SCA1.


Subject(s)
Ataxin-1/drug effects , Neurodegenerative Diseases/genetics , Oligonucleotides, Antisense/therapeutic use , Spinocerebellar Ataxias/classification , Animals , Ataxin-1/metabolism , Female , Magnetic Resonance Spectroscopy/methods , Male , Mice , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/drug therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/adverse effects , Phenotype , Sequence Analysis, RNA/methods , Spinocerebellar Ataxias/diagnostic imaging , Spinocerebellar Ataxias/drug therapy , Spinocerebellar Ataxias/genetics , Survival Analysis , Transcriptome
10.
Neurobiol Dis ; 116: 93-105, 2018 08.
Article in English | MEDLINE | ID: mdl-29758256

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a polyglutamine (polyQ) repeat neurodegenerative disease in which a primary site of pathogenesis are cerebellar Purkinje cells. In addition to polyQ expansion of ataxin-1 protein (ATXN1), phosphorylation of ATXN1 at the serine 776 residue (ATXN1-pS776) plays a significant role in protein toxicity. Utilizing a biochemical approach, pharmacological agents and cell-based assays, including SCA1 patient iPSC-derived neurons, we examine the role of Protein Kinase A (PKA) as an effector of ATXN1-S776 phosphorylation. We further examine the implications of PKA-mediated phosphorylation at ATXN1-S776 on SCA1 through genetic manipulation of the PKA catalytic subunit Cα in Pcp2-ATXN1[82Q] mice. Here we show that pharmacologic inhibition of S776 phosphorylation in transfected cells and SCA1 patient iPSC-derived neuronal cells lead to a decrease in ATXN1. In vivo, reduction of PKA-mediated ATXN1-pS776 results in enhanced degradation of ATXN1 and improved cerebellar-dependent motor performance. These results provide evidence that PKA is a biologically important kinase for ATXN1-pS776 in cerebellar Purkinje cells.


Subject(s)
Ataxia/metabolism , Ataxin-1/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Purkinje Cells/metabolism , Serine/metabolism , Animals , Ataxia/genetics , Ataxia/pathology , Ataxin-1/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Female , Humans , Male , Mice , Mice, Transgenic , Phosphorylation/physiology , Purkinje Cells/pathology , Serine/genetics
11.
Neuron ; 97(6): 1235-1243.e5, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29526553

ABSTRACT

Polyglutamine (polyQ) diseases are caused by expansion of translated CAG repeats in distinct genes leading to altered protein function. In spinocerebellar ataxia type 1 (SCA1), a gain of function of polyQ-expanded ataxin-1 (ATXN1) contributes to cerebellar pathology. The extent to which cerebellar toxicity depends on its cognate partner capicua (CIC), versus other interactors, remains unclear. It is also not established whether loss of the ATXN1-CIC complex in the cerebellum contributes to disease pathogenesis. In this study, we exclusively disrupt the ATXN1-CIC interaction in vivo and show that it is at the crux of cerebellar toxicity in SCA1. Importantly, loss of CIC in the cerebellum does not cause ataxia or Purkinje cell degeneration. Expression profiling of these gain- and loss-of-function models, coupled with data from iPSC-derived neurons from SCA1 patients, supports a mechanism in which gain of function of the ATXN1-CIC complex is the major driver of toxicity.


Subject(s)
Ataxin-1/deficiency , Cerebellum/metabolism , Gain of Function Mutation/physiology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism , Animals , Ataxin-1/genetics , Cells, Cultured , Cerebellum/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spinocerebellar Ataxias/pathology
12.
Neuron ; 89(6): 1194-1207, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26948890

ABSTRACT

SCA1, a fatal neurodegenerative disorder, is caused by a CAG expansion encoding a polyglutamine stretch in the protein ATXN1. We used RNA sequencing to profile cerebellar gene expression in Pcp2-ATXN1[82Q] mice with ataxia and progressive pathology and Pcp2-ATXN1[30Q]D776 animals having ataxia in absence of Purkinje cell progressive pathology. Weighted Gene Coexpression Network Analysis of the cerebellar expression data revealed two gene networks that significantly correlated with disease and have an expression profile correlating with disease progression in ATXN1[82Q] Purkinje cells. The Magenta Module provides a signature of suppressed transcriptional programs reflecting disease progression in Purkinje cells, while the Lt Yellow Module reflects transcriptional programs activated in response to disease in Purkinje cells as well as other cerebellar cell types. Furthermore, we found that upregulation of cholecystokinin (Cck) and subsequent interaction with the Cck1 receptor likely underlies the lack of progressive Purkinje cell pathology in Pcp2-ATXN1[30Q]D776 mice.


Subject(s)
Ataxin-1/genetics , Cerebellum/metabolism , Cerebellum/pathology , Spinocerebellar Ataxias/pathology , Transcriptome/genetics , Animals , Ataxin-1/metabolism , Chemokines, CC/deficiency , Chemokines, CC/genetics , Cholecystokinin/deficiency , Cholecystokinin/genetics , Disease Models, Animal , Disease Progression , Gene Regulatory Networks , Guanine Nucleotide Exchange Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Purkinje Cells/metabolism , Receptor, Cholecystokinin B/deficiency , Receptor, Cholecystokinin B/genetics , Up-Regulation/genetics
13.
Nature ; 498(7454): 325-331, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23719381

ABSTRACT

Many neurodegenerative disorders, such as Alzheimer's, Parkinson's and polyglutamine diseases, share a common pathogenic mechanism: the abnormal accumulation of disease-causing proteins, due to either the mutant protein's resistance to degradation or overexpression of the wild-type protein. We have developed a strategy to identify therapeutic entry points for such neurodegenerative disorders by screening for genetic networks that influence the levels of disease-driving proteins. We applied this approach, which integrates parallel cell-based and Drosophila genetic screens, to spinocerebellar ataxia type 1 (SCA1), a disease caused by expansion of a polyglutamine tract in ataxin 1 (ATXN1). Our approach revealed that downregulation of several components of the RAS-MAPK-MSK1 pathway decreases ATXN1 levels and suppresses neurodegeneration in Drosophila and mice. Importantly, pharmacological inhibitors of components of this pathway also decrease ATXN1 levels, suggesting that these components represent new therapeutic targets in mitigating SCA1. Collectively, these data reveal new therapeutic entry points for SCA1 and provide a proof-of-principle for tackling other classes of intractable neurodegenerative diseases.


Subject(s)
Drosophila melanogaster/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/toxicity , Nuclear Proteins/metabolism , Nuclear Proteins/toxicity , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology , ras Proteins/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Ataxin-1 , Ataxins , Cell Line, Tumor , Disease Models, Animal , Down-Regulation/drug effects , Drosophila melanogaster/genetics , Female , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Molecular Sequence Data , Molecular Targeted Therapy , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphorylation , Protein Stability/drug effects , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Transgenes
14.
J Neurosci ; 33(13): 5806-20, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23536093

ABSTRACT

Previous studies indicate that while transgenic mice with ATXN1[30Q]-D776-induced disease share pathological features caused by ATXN1[82Q] having an expanded polyglutamine tract, they fail to manifest the age-related progressive neurodegeneration seen in spinocerebellar ataxia type 1. The shared features include morphological alterations in climbing fiber (CF) innervation of Purkinje cells (PCs). To further investigate the ability of ataxin-1 (ATXN1) to impact CF/PC innervation, this study used morphological and functional approaches to examine CF/PC innervation during postnatal development in ATXN1[30Q]-D776 and ATXN1[82Q] cerebella. Notably, ATXN1[30Q]-D776 induced morphological alterations consistent with the development of the innervation of PCs by CFs being compromised, including a reduction of CF translocation along the PC dendritic tree, and decreased pruning of CF terminals from the PC soma. As previously shown for ATXN1[82Q], ATXN1[30Q]-D776 must enter the nucleus of PCs to induce these alterations. Experiments using conditional ATXN1[30Q]-D776 mice demonstrate that both the levels and specific timing of mutant ATXN1 expression are critical for alteration of the CF-PC synapse. Together these observations suggest that ATXN1, expressed exclusively in PCs, alters expression of a gene(s) in the postsynaptic PC that are critical for its innervation by CFs. To investigate whether ATXN1[30Q]-D776 curbs the progressive disease in ATXN1[82Q]-S776 mice, we crossed ATXN1[30Q]-D776 and ATXN1[82Q]-S776 mice and found that double transgenic mice developed progressive PC atrophy. Thus, the results also show that to develop progressive cerebellar degeneration requires expressing ATXN1 with an expanded polyglutamine tract.


Subject(s)
Cerebellum/growth & development , Cerebellum/pathology , Nerve Fibers/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Purkinje Cells/metabolism , Spinocerebellar Ataxias/pathology , Synapses/pathology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Ataxin-1 , Ataxins , Calbindins , Disability Evaluation , Disease Models, Animal , Electric Stimulation , Fluorescent Dyes , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Humans , Membrane Potentials/genetics , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Mutation/genetics , Nerve Fibers/metabolism , Nerve Fibers/physiology , Nerve Tissue Proteins/genetics , Neuropeptides/genetics , Nuclear Proteins/genetics , Optical Imaging , Patch-Clamp Techniques , RNA, Messenger/metabolism , S100 Calcium Binding Protein G/metabolism , Spinocerebellar Ataxias/genetics , Synapses/genetics , Vesicular Glutamate Transport Protein 2/metabolism
15.
J Neurosci ; 31(36): 12778-89, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21900557

ABSTRACT

One fundamental unanswered question in the field of polyglutamine diseases concerns the pathophysiology of neuronal dysfunction. Is there dysfunction in a specific neuronal population or circuit initially that contributes the onset of behavioral abnormalities? This study used a systems-level approach to investigate the functional integrity of the excitatory cerebellar cortical circuitry in vivo from several transgenic ATXN1 mouse lines. We tested the hypotheses that there are functional climbing fiber (CF)-Purkinje cell (PC) and parallel fiber (PF)-PC circuit abnormalities using flavoprotein autofluorescence optical imaging and extracellular field potential recordings. In early-symptomatic and symptomatic animals expressing ATXN1[82Q], there is a marked reduction in PC responsiveness to CF activation. Immunostaining of vesicular glutamate transporter type 2 demonstrated a decrement in CF extension on PC dendrites in symptomatic ATXN1[82Q] mice. In contrast, responses to PF stimulation were relatively normal. Importantly, the deficits in CF-PC synaptic transmission required expression of pathogenic ataxin-1 (ATXN1[82Q]) and for its entrance into the nucleus of PCs. Loss of endogenous mouse Atxn1 had no discernible effects. Furthermore, the abnormalities in CF-PC synaptic transmission were ameliorated when mutant transgene expression was prevented during postnatal cerebellar development. The results demonstrate the preferential susceptibility of the CF-PC circuit to the effects of ATXN1[82Q]. Further, this deficit likely contributes to the abnormal motor phenotype of ATXN1[82Q] mice. For polyglutamine diseases generally, the findings support a model whereby specific neuronal circuits suffer insults that alter function before cell death.


Subject(s)
Nerve Fibers/pathology , Nerve Tissue Proteins/genetics , Neural Pathways/pathology , Neurons/pathology , Nuclear Proteins/genetics , Purkinje Cells/pathology , Spinocerebellar Ataxias/pathology , Animals , Ataxin-1 , Ataxins , Blotting, Western , Cell Death/physiology , Electrophysiological Phenomena , Female , Immunohistochemistry , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Movement Disorders/genetics , Movement Disorders/pathology , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Patch-Clamp Techniques , Spinocerebellar Ataxias/genetics , Synaptic Transmission/genetics , Synaptic Transmission/physiology
16.
Hum Mol Genet ; 20(11): 2204-12, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21427130

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is one of nine dominantly inherited neurodegenerative diseases caused by polyglutamine tract expansion. In SCA1, the expanded polyglutamine tract is in the ataxin-1 (ATXN1) protein. ATXN1 is part of an in vivo complex with retinoid acid receptor-related orphan receptor alpha (Rora) and the acetyltransferase tat-interactive protein 60 kDa (Tip60). ATXN1 and Tip60 interact directly via the ATXN1 and HMG-box protein 1 (AXH) domain of ATXN1. Moreover, the phospho-mimicking Asp amino acid at position 776, previously shown to enhance pathogenesis, increases the ability of ATXN1 to interact with Tip60. Using a genetic approach, the biological relevance of the ATXN1/Tip60 interaction was assessed by crossing ATXN1[82Q] mice with Tip60(+/-)animals. Partial Tip60 loss increased Rora and Rora-mediated gene expression and delayed ATXN1[82]-mediated cerebellar degeneration during mid-stage disease progression. These results suggested a specific, temporal role for Tip60 during disease progression. We also showed that genetic background modulated ATXN1[82Q]-induced phenotypes. Of interest, these latter studies showed that some phenotypes are enhanced on a mixed background while others are suppressed.


Subject(s)
Histone Acetyltransferases/genetics , Nerve Degeneration/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Spinocerebellar Ataxias/genetics , Animals , Ataxin-1 , Ataxins , CHO Cells , Chromosome Mapping , Cricetinae , Cricetulus , Disease Models, Animal , Gene Expression Regulation , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Haploinsufficiency , Histone Acetyltransferases/metabolism , Lysine Acetyltransferase 5 , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Peptides/genetics , Phenotype , Purkinje Cells/metabolism , Purkinje Cells/pathology , Trans-Activators
17.
Neuron ; 67(6): 929-35, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20869591

ABSTRACT

Glutamine tract expansion triggers nine neurodegenerative diseases by conferring toxic properties to the mutant protein. In SCA1, phosphorylation of ATXN1 at Ser776 is thought to be key for pathogenesis. Here, we show that replacing Ser776 with a phosphomimicking Asp converted ATXN1 with a wild-type glutamine tract into a pathogenic protein. ATXN1[30Q]-D776-induced disease in Purkinje cells shared most features with disease caused by ATXN1[82Q] having an expanded polyglutamine tract. However, in contrast to disease induced by ATXN1[82Q] that progresses to cell death, ATXN1[30Q]-D776 failed to induce cell death. These results support a model where pathogenesis involves changes in regions of the protein in addition to the polyglutamine tract. Moreover, disease initiation and progression to neuronal dysfunction are distinct from induction of cell death. Ser776 is critical for the pathway to neuronal dysfunction, while an expanded polyglutamine tract is essential for neuronal death.


Subject(s)
Aspartic Acid/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Serine/genetics , Spinocerebellar Ataxias/genetics , Animals , Ataxin-1 , Ataxins , Calbindins , Cerebellum/pathology , Dendrites/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Mice , Mice, Transgenic , Motor Activity/genetics , Neural Pathways/metabolism , Neural Pathways/pathology , Purkinje Cells/pathology , Purkinje Cells/ultrastructure , Rotarod Performance Test , S100 Calcium Binding Protein G/metabolism , Spinocerebellar Ataxias/pathology , Spinocerebellar Ataxias/physiopathology , Vesicular Glutamate Transport Protein 2/metabolism
18.
J Neurochem ; 110(2): 675-86, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19500214

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is one of nine inherited neurodegenerative disorders caused by a mutant protein with an expanded polyglutamine tract. Phosphorylation of ataxin-1 (ATXN1) at serine 776 is implicated in SCA1 pathogenesis. Previous studies, utilizing transfected cell lines and a Drosophila photoreceptor model of SCA1, suggest that phosphorylating ATXN1 at S776 renders it less susceptible to degradation. This work also indicated that oncogene from AKR mouse thymoma (Akt) promotes the phosphorylation of ATXN1 at S776 and severity of neurodegeneration. Here, we examined the phosphorylation of ATXN1 at S776 in cerebellar Purkinje cells, a prominent site of pathology in SCA1. We found that while phosphorylation of S776 is associated with a stabilization of ATXN1 in Purkinje cells, inhibition of Akt either in vivo or in a cerebellar extract-based phosphorylation assay did not decrease the phosphorylation of ATXN1-S776. In contrast, immunodepletion and inhibition of cyclic AMP-dependent protein kinase decreased phosphorylation of ATXN1-S776. These results argue against Akt as the in vivo kinase that phosphorylates S776 of ATXN1 and suggest that cyclic AMP-dependent protein kinase is the active ATXN1-S776 kinase in the cerebellum.


Subject(s)
Cerebellum/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Serine/metabolism , Alanine/genetics , Alanine/metabolism , Amino Acid Sequence , Animals , Ataxin-1 , Ataxins , Cerebellum/enzymology , Enzyme Stability/genetics , Humans , Mice , Mice, Inbred AKR , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Phosphorylation , Point Mutation , Proto-Oncogene Proteins c-akt/genetics , Purkinje Cells/enzymology , Purkinje Cells/metabolism , Serine/genetics
19.
Cell ; 127(7): 1335-47, 2006 Dec 29.
Article in English | MEDLINE | ID: mdl-17190598

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is one of several neurodegenerative diseases caused by expansion of a polyglutamine tract in the disease protein, in this case, ATAXIN-1 (ATXN1). A key question in the field is whether neurotoxicity is mediated by aberrant, novel interactions with the expanded protein or whether its wild-type functions are augmented to a deleterious degree. We examined soluble protein complexes from mouse cerebellum and found that the majority of wild-type and expanded ATXN1 assembles into large stable complexes containing the transcriptional repressor Capicua. ATXN1 directly binds Capicua and modulates Capicua repressor activity in Drosophila and mammalian cells, and its loss decreases the steady-state level of Capicua. Interestingly, the S776A mutation, which abrogates the neurotoxicity of expanded ATXN1, substantially reduces the association of mutant ATXN1 with Capicua in vivo. These data provide insight into the function of ATXN1 and suggest that SCA1 neuropathology depends on native, not novel, protein interactions.


Subject(s)
Cerebellum/metabolism , Drosophila/physiology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Spinocerebellar Ataxias/etiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Ataxin-1 , Ataxins , Brain/metabolism , Conserved Sequence , Drosophila/embryology , Eye Abnormalities/etiology , Humans , Mice , Molecular Sequence Data , Mutation , Peptides/metabolism , Sequence Homology, Amino Acid , Spinocerebellar Ataxias/genetics , Transcription, Genetic , Wings, Animal/abnormalities
20.
Cell ; 127(4): 697-708, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-17110330

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is one of nine inherited, typically adult onset, polyglutamine neurodegenerative diseases. To examine whether development impacts SCA1, we used a conditional transgenic mouse model of SCA1 to delay the postnatal expression of mutant ATXN1 until after completion of cerebellar development. Delayed postnatal expression of mutant ATXN1 led to a substantial reduction in severity of disease in adults in comparison with early postnatal gene expression. This was linked to a destabilization of RORalpha, a transcription factor critical for cerebellar development. In SCA1 mice, there was a depletion of RORalpha and a reduction in expression of genes controlled by RORalpha. Partial loss of RORalpha enhanced mutant ATXN1 pathogenicity. Additionally, evidence points to the existence of a complex containing ATXN1, RORalpha, and the RORalpha coactivator Tip60. These studies indicate RORalpha and Tip60 have a role in SCA1 and suggest a mechanism by which compromising cerebellar development contributes to severity of neurodegeneration in an adult.


Subject(s)
Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Purkinje Cells/cytology , Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/metabolism , Animals , Ataxin-1 , Ataxins , COS Cells , Chlorocebus aethiops , Disease Progression , Down-Regulation/genetics , Histone Acetyltransferases/metabolism , Humans , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Mutant Proteins/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1 , Protein Binding , Protein Interaction Mapping , Purkinje Cells/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Trans-Activators/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...